1
|
Dastmalchi N, Alipour MR, Safaralizadeh R, Hajiasgharzadeh K. An Updated Review on Dysregulated lncRNAs and their Contribution to the Various Molecular Types of Lung Carcinoma. Anticancer Agents Med Chem 2025; 25:490-498. [PMID: 39754779 DOI: 10.2174/0118715206336608241104065557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 01/06/2025]
Abstract
Lung cancer is correlated with a high death rate, with approximately 1.8 million mortality cases reported worldwide in 2022. Despite development in the control of lung cancer, most cases are detected at higher stages with short survival rates. This reveals a need to recognize novel techniques to treat malignancy and decrease the burden of lung cancer. Long noncoding RNAs (lncRNAs) manage vital cellular and biochemical functions. lncRNAs play crucial roles in transcriptional and translational processes and signaling cascades. Recently, lncRNAs have been reported to be associated with malignancy where their expression is deregulated, leading to abnormal cellular activities and signaling pathways. In various malignancies, including lung cancer, lncRNA deregulation disrupts normal cellular function, promoting tumorigenesis and influencing patient outcomes and treatment responses. Studies have shown that lncRNAs can act as both oncogenes and tumor suppressors, depending on the lung cancer subtype, specifically in Non-small Cell Lung Cancer (NSCLC) and Small Cell Lung Cancer (SCLC). This dual role of lncRNAs as critical biomarkers might provide insights into lung cancer development and progression. lncRNAs have been discussed as key biomarkers in lung cancer. A comprehensive understanding of the biological activities of lncRNAs in NSCLC and SCLC may improve prognosis, diagnosis, and therapeutic methods. Researchers are increasingly interested in lncRNAs as potential diagnostic biomarkers and therapeutic targets in cancer treatment. As researchers continue to explore lncRNAs, their pivotal roles in lung cancer become increasingly evident. This review highlights the function of lncRNAs in lung carcinogenesis and discusses their molecular mechanisms of function.
Collapse
Affiliation(s)
- Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | | | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
2
|
Saraswat SK, Mahmood BS, Ajila F, Kareem DS, Alwan M, Athab ZH, Shaier JB, Hosseinifard SR. Deciphering the oncogenic landscape: Unveiling the molecular machinery and clinical significance of LncRNA TMPO-AS1 in human cancers. Pathol Res Pract 2024; 255:155190. [PMID: 38330619 DOI: 10.1016/j.prp.2024.155190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024]
Abstract
The in-depth exploration of long non-coding RNAs (lncRNAs) reveals their pivotal and diverse roles in various disorders, particularly cancer. Within this intricate landscape, thymopoietin-antisense RNA-1 (TMPO-AS1) emerges as a noteworthy instigator of oncogenesis in humans. This exhaustive review seeks to intricately unravel the present understanding of TMPO-AS1, emphasizing its molecular foundations and highlighting its clinical applications in the realm of cancer research. TMPO-AS1 consistently exhibits heightened expression across a spectrum of cancer types, encompassing lung, colorectal, breast, cervical, bladder, pancreatic, hepatocellular, gastric, ovarian, and osteosarcoma. Elevated levels of TMPO-AS1 are intricately linked to unfavorable prognoses, accompanied by distinctive clinical and pathological characteristics. Functionally, TMPO-AS1 showcases its prowess in enhancing cancer cell migration, invasion, proliferation, and orchestrating epithelial-mesenchymal transition (EMT) through a myriad of molecular mechanisms. These mechanisms entail intricate interactions with proteins, microRNAs, and intricate signaling pathways. Furthermore, TMPO-AS1 is intricately involved in regulating critical cellular processes, including apoptosis and the cell cycle. The mounting evidence converges towards the potential of TMPO-AS1 serving as a diagnostic and prognostic biomarker, further entwined with its potential role in influencing chemoresistance in cancer. This potential is underscored by its consistent associations with clinical outcomes and treatment responses. This comprehensive investigation not only consolidates our existing knowledge of TMPO-AS1's multifaceted roles but also sheds illuminating insights on its profound significance in the intricate landscape of cancer biology, paving the way for potential applications in clinical practice.
Collapse
Affiliation(s)
| | | | - Freddy Ajila
- Facultad de Informática y Electrónica, Escuela Superior Politécnica de Chimborazo (ESPOCH), Sede Orellana, El Coca 220001, Ecuador.
| | | | - Mariem Alwan
- Medical Technical College, Al-Farahidi University, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | | | |
Collapse
|
3
|
Jin Y, Jiang A, Sun L, Lu Y. Long noncoding RNA TMPO-AS1 accelerates glycolysis by regulating the miR-1270/PKM2 axis in colorectal cancer. BMC Cancer 2024; 24:238. [PMID: 38383342 PMCID: PMC10880273 DOI: 10.1186/s12885-024-11964-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Long noncoding RNA thymopoietin-antisense RNA 1 (TMPO-AS1) is recognized as a participant in cancer progression. Nevertheless, its biological function in colorectal cancer remains obscure and needs further elucidation. METHODS AND RESULTS First, we discovered enriched TMPO-AS1 in the tumor tissues that were related to poor prognosis. TMPO-AS1 knockdown enhanced SW480 cell apoptosis but inhibited invasion, proliferation, migration, and glucose metabolism. Further, MiR-1270 is directly bound with TMPO-AS1. MiR-1270 mimics were confirmed to inhibit cell proliferation, invasion, and glucose metabolism in our study. Mechanistically, miR-1270 directly is bound with the 3' untranslated regions (3'UTR) of PKM2 to downregulate PKM2. MiR-1270 inhibitors reversed the TMPO-AS1 knockdown's effect on suppressing the tumor cell proliferation, invasion, and glycolysis, while the knockdown of PKM2 further inverted the function of miR-1270 inhibitors on the TMPO-AS1 knockdown. CONCLUSIONS This study illustrated that TMPO-AS1 advanced the development and the glycolysis of colorectal cancer by modulating the miR-1270/PKM2 axis, which provided a new insight into the colorectal cancer therapeutic strategy.
Collapse
Affiliation(s)
- Yingmin Jin
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China.
| | - Aimin Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| | - Liying Sun
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| | - Yue Lu
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Str, Harbin, 150001, People's Republic of China
| |
Collapse
|
4
|
Zabeti Touchaei A, Vahidi S, Samadani AA. Decoding the regulatory landscape of lncRNAs as potential diagnostic and prognostic biomarkers for gastric and colorectal cancers. Clin Exp Med 2024; 24:29. [PMID: 38294554 PMCID: PMC10830721 DOI: 10.1007/s10238-023-01260-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/12/2023] [Indexed: 02/01/2024]
Abstract
Colorectal cancer (CRC) and gastric cancer (GC) are major contributors to cancer-related mortality worldwide. Despite advancements in understanding molecular mechanisms and improved drug treatments, the overall survival rate for patients remains unsatisfactory. Metastasis and drug resistance are major challenges contributing to the high mortality rate in both CRC and GC. Recent research has shed light on the role of long noncoding RNAs (lncRNAs) in the development and progression of these cancers. LncRNAs regulate gene expression through various mechanisms, including epigenetic modifications and interactions with microRNAs (miRNAs) and proteins. They can serve as miRNA precursors or pseudogenes, modulating gene expression at transcriptional and post-transcriptional levels. Additionally, circulating lncRNAs have emerged as non-invasive biomarkers for the diagnosis, prognosis, and prediction of drug therapy response in CRC and GC. This review explores the intricate relationship between lncRNAs and CRC/GC, encompassing their roles in cancer development, progression, and chemoresistance. Furthermore, it discusses the potential of lncRNAs as therapeutic targets in these malignancies. The interplay between lncRNAs, miRNAs, and tumor microenvironment is also highlighted, emphasizing their impact on the complexity of cancer biology. Understanding the regulatory landscape and molecular mechanisms governed by lncRNAs in CRC and GC is crucial for the development of effective diagnostic and prognostic biomarkers, as well as novel therapeutic strategies. This review provides a comprehensive overview of the current knowledge and paves the way for further exploration of lncRNAs as key players in the management of CRC and GC.
Collapse
Affiliation(s)
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
5
|
Ghafouri-Fard S, Askari A, Hussen BM, Taheri M, Mokhtari M. A long non-coding RNA with important roles in the carcinogenesis. Front Cell Dev Biol 2022; 10:1037149. [PMID: 36467407 PMCID: PMC9709449 DOI: 10.3389/fcell.2022.1037149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2023] Open
Abstract
Long non-coding RNAs are demonstrated to contribute to carcinogenesis. TMPO Antisense RNA 1 (TMPO-AS1) is an example of lncRNAs with crucial roles in this process. This lncRNA serves as a sponge for miR-320a, miR-383-5p, miR-329-3p, miR-126, miR-329, miR-199a-5p, miR-577, miR-4731-5p, miR-140-5p, miR-1179, miR-143-3p, miR-326, miR-383-5p, let-7c-5p, let-7g-5p, miR-199a-5p, miR-200c, miR-204-3p, miR-126-5p, miR-383-5p, miR-498, miR-143-3p, miR-98-5p, miR-140 and miR-143. It can also affect activity of PI3K/Akt/mTOR pathway. The current review summarizes the role of TMPO-AS1 in the carcinogenesis and assessment of its potential as a marker for certain types of cancers.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arian Askari
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Majid Mokhtari
- Skull Base Research Center, Loghman Hakam Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Ye J, Yan Y, Xin L, Liu J, Tang T, Bao X. Long non-coding RNA TMPO-AS1 facilitates the progression of colorectal cancer cells via sponging miR-98-5p to upregulate BCAT1 expression. J Gastroenterol Hepatol 2022; 37:144-153. [PMID: 34370878 DOI: 10.1111/jgh.15657] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/09/2021] [Accepted: 08/03/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIM Colorectal cancer, as a common malignant carcinoma in the gastrointestinal tract, has a high mortality globally. However, the specific molecular mechanisms of long non-coding RNA (lncRNA) thymopoietin antisense transcript 1 (TMPO-AS1) in colorectal cancer were unclear. METHODS We tested the expression level of TMPO-AS1 via qRT-PCR in colorectal cancer cells, while the protein levels of branched chain amino acid transaminase 1 (BCAT1) and the stemness-related proteins were evaluated by western blot analysis. Colony formation, EdU staining, TUNEL, flow cytometry, and sphere formation assays were to assess the biological behaviors of colorectal cancer cells. Then, luciferase reporter, RIP, and RNA pull down assay were applied for confirming the combination between microRNA-98-5p (miR-98-5p) and TMPO-AS1/BCAT1. RESULTS TMPO-AS1 was aberrantly expressed at high levels in colorectal cancer cells. Silenced TMPO-AS1 restrained cell proliferation and stemness and promoted apoptosis oppositely, while overexpressing TMPO-AS1 exerted the adverse effects. Furthermore, miR-98-5p was proven to a target of TMPO-AS1 inhibit cell progression in colorectal cancer. Additionally, BCAT1 was proved to enhance cell progression as the target of miR-98-5p, and it offset the effect of silenced TMPO-AS1 on colorectal cancer cells. CONCLUSION TMPO-AS1 promotes the progression of colorectal cancer cells via sponging miR-98-5p to upregulate BCAT1 expression.
Collapse
Affiliation(s)
- Jinjun Ye
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| | - Yukuang Yan
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| | - Le Xin
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| | - Jidong Liu
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| | - Tao Tang
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| | - Xing Bao
- Department of General Surgery, Longgang Central Hospital, Shenzhen, China
| |
Collapse
|
7
|
Ghodrati R, Safaralizadeh R, Dastmalchi N, Hosseinpourfeizi M, Asadi M, Shirmohammadi M, Baradaran B. Overexpression of lncRNA DLEU1 in Gastric Cancer Tissues Compared to Adjacent Non-Tumor Tissues. J Gastrointest Cancer 2021; 53:990-994. [PMID: 34738190 DOI: 10.1007/s12029-021-00733-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Gastric cancer (GC) is caused by environmental factors and genetic changes of protein-coding- and non-coding sequences, which entail short non-coding RNAs (microRNAs) and long non-coding RNAs (lncRNAs). DLEU1 (deleted in lymphocytic leukemia 1), as an effective lncRNA located on chromosome 14.3q 13, modulates the nuclear factor-kB (NF-kB) signaling pathway. This gene usually plays an oncogenic role in the tumorigenesis of multiple types of cancer. The present study examined the expression level of DLEU1 and its association with clinical-pathological characteristics in GC. METHODS Total RNA of 100 specimens was extracted by TRIzol reagent. After cDNA synthesis, qRT-PCR analysis was performed to measure the expression level of the DLEU1 gene and the obtained data were analyzed by SPSS 16.0. RESULTS The relative expression level of DLEU1 significantly increased in tumor specimens compared to the normal tumor margin specimens. The biomarker index of lncRNA DLEU1 was 0.7 in tumor tissues. The observed high expression level of DLEU1 was pertinent to the pathological progressive TNM stage, lymph node metastasis, differentiation degree, patient's age and lifestyle, and Helicobacter pylori infection in GC patients. CONCLUSION The obtained findings suggested that DLEU1 acts as an oncogene in GC and might be a new target for gene therapy of GC.
Collapse
Affiliation(s)
- Roghieh Ghodrati
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | | | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Shirmohammadi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Lu H, Zhang Z, Lu Y, Xiu W, Cui J. LncRNA NEAT1 Acts as an miR-148b-3p Sponge to Regulate ROCK1 Inhibition of Retinoblastoma Growth. Cancer Manag Res 2021; 13:5587-5597. [PMID: 34285579 PMCID: PMC8285126 DOI: 10.2147/cmar.s271326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/15/2020] [Indexed: 01/26/2023] Open
Abstract
Background It is reported that long non-coding RNA nuclear paraspeckle assembly transcript 1 (LncRNA NEAT1) is involved in the occurrence and development of various cancers. However, the detailed biological function and mechanism of LncRNA NEAT1 in retinoblastoma are still unclear. So we will explore the biological function and possible mechanism of LncRNA NEAT1 in retinoblastoma. Materials and Methods Quantitative real-time PCR (qRT-PCR) was used to detect LncRNA NEAT1 in retinoblastoma tissues and cell lines. Cell counting kit 8, Transwell and flow cytometry were applied to explore cell proliferation, invasion and apoptosis. The target miRNAs (miR) of LncRNA NEAT1 and miR and downstream target genes were predicted using Starbase3.0 software and confirmed by double luciferase reporting test and RNA binding protein immunoprecipitation (RIP). Western Blot was applied to explore ROCK1 in cells, and tumor allogeneic experiment was applied to study the role of LncRNA NEAT1 on tumor growth. Results It was found that LncRNA NEAT1 was up-regulated in retinoblastoma tissues, cells and serum, and the prognosis of patients with high expression of LNC RNA NEAT 1 was poor. Functional analysis showed that knocking down LncRNA NEAT1 could weaken proliferation and invasion, and accelerate apoptosis. Tumor allogeneic experiment showed that sh-NEAT1 injection can inhibit tumor growth. In addition, LncRNA NEAT1 inhibited proliferation and invasion, and promoted apoptosis through miR-148b-3p/ROCK1 axis. Conclusion LncRNA NEAT1 can mediate miR-148b-3p/ROCK1 axis to weaken the proliferation and invasion of retinoblastoma.
Collapse
Affiliation(s)
- Hang Lu
- Research Center of Ophthalmology, The First Hospital of Qiqihar, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang Province, People's Republic of China
| | - Zhenjun Zhang
- Ophthalmology Department, Beiman Hongpeng Hospital of Qiqihar, Qiqihar, Heilongjiang Province, People's Republic of China
| | - Yao Lu
- International Education College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, People's Republic of China
| | - Weiwei Xiu
- Research Center of Ophthalmology, The First Hospital of Qiqihar, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang Province, People's Republic of China
| | - Jinglin Cui
- Research Center of Ophthalmology, The First Hospital of Qiqihar, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, Heilongjiang Province, People's Republic of China
| |
Collapse
|
9
|
Hu Y, Zhang Y, Ding M, Xu R. Long noncoding RNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol 2021; 36:1877-1888. [PMID: 33295056 DOI: 10.1111/jgh.15362] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/30/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIM Gastric cancer (GC) is an aggressive tumor featured by uncontrolled cell proliferation and metastasis. In recent years, long noncoding RNAs (lncRNAs) act as crucial regulators and biological markers in multiple cancers. LncRNA TMPO-AS1 has been revealed to be an oncogene in some cancers. Nevertheless, there is little known about the biological role of TMPO-AS1 in GC. METHODS Reverse transcription-quantitative polymerase chain reaction analysis was used to examine the expression level of TMPO-AS1 in GC tissues and cells. Cell Counting Kit-8, colony formation, wound healing assays, and western blot analysis were performed to determine the role of TMPO-AS1 in GC cells. RNA pull-down, luciferase reporter, and RNA immunoprecipitation assays were used to test the interaction among TMPO-AS1, miR-126-5p, and BRCC3. RESULTS TMPO-AS1 was highly expressed in GC tissues and cells. Upregulated TMPO-AS1 was closely associated with adverse prognosis of GC patients. Functional assays showed that TMPO-AS1 promoted GC cell proliferation, migration, and angiogenesis. Furthermore, it was found that TMPO-AS1 acted as a competing endogenous RNA for miR-126-5p to upregulate BRCC3 expression. Rescue assays revealed that TMPO-AS1 facilitated cellular progression of GC by sponging miR-126-5p and upregulating BRCC3. In addition, we found that the effects of the TMPO-AS1/miR-126-5p/BRCC3 axis on GC cell progression were related to the PI3K/Akt/mTOR pathway. CONCLUSIONS Our study demonstrated that the TMPO-AS1/miR-126-5p/BRCC3 axis was involved in GC progression via the regulation of PI3K/Akt/mTOR pathway, which might provide a potential therapeutic strategy for GC.
Collapse
Affiliation(s)
- Yu Hu
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Ding
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ruisi Xu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Zheng Q, Jia J, Zhou Z, Chu Q, Lian W, Chen Z. The Emerging Role of Thymopoietin-Antisense RNA 1 as Long Noncoding RNA in the Pathogenesis of Human Cancers. DNA Cell Biol 2021; 40:848-857. [PMID: 34096793 DOI: 10.1089/dna.2021.0024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play essential roles in the occurrence and development of multiple human cancers. An accumulating body of researches have investigated thymopoietin antisense RNA 1 (TMPO-AS1) as a newly discovered lncRNA, which functions as an oncogenic lncRNA that is upregulated in various human malignancies and associated with poor prognosis. Many studies have detected abnormally high expression levels of TMPO-AS1 in multiple cancers, such as lung cancer, breast cancer, colorectal cancer (CRC), hepatocellular carcinoma, CRC, gastric cancer, ovarian cancer, thyroid cancer, esophageal cancer, Wilms tumor, cervical cancer, retinoblastoma, bladder cancer, osteosarcoma, and prostate cancer. TMPO-AS1 has been subsequently demonstrated to play a pivotal role in tumorigenesis and progression. The aberrantly expressed TMPO-AS1 acts as a competing endogenous RNA (ceRNA) that inhibits miRNA expression, thus activating the expression of downstream oncogenes. This study comprehensively summarizes the aberrant expressions of TMPO-AS1 as reported in the current literature and explains the relevant biological regulation mechanisms in carcinogenesis and tumor progression. Corresponding studies have indicated that TMPO-AS1 has a potential value as a promising biomarker or a target for cancer therapy.
Collapse
Affiliation(s)
- Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Junjun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziyuan Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Lian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Xue F, Song X, Zhang S, Niu M, Cui Y, Wang Y, Zhao T. Long non-coding RNA TMPO-AS1 serves as a tumor promoter in pancreatic carcinoma by regulating miR-383-5p/SOX11. Oncol Lett 2021; 21:255. [PMID: 33664819 PMCID: PMC7882873 DOI: 10.3892/ol.2021.12517] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
The dysregulation of lncRNA TMPO antisense RNA 1 (TMPO-AS1) has been detected in various malignant tumors. However, the role of lncRNA TMPO-AS1 remains unclear in pancreatic carcinoma. The present study aimed to elucidate the functional mechanism of TMPO-AS1 in pancreatic carcinoma. In the present study, RT-qPCR, western blotting, MTT, Transwell, luciferase reporter and xenograft assays were used to investigate the role of lncRNA TMPO-AS1 in pancreatic carcinoma. Upregulation of lncRNA TMPO-AS1 was revealed in pancreatic carcinoma tissues and cells. Furthermore, knockdown of TMPO-AS1 restrained cell proliferation and motility in pancreatic carcinoma. In addition, microRNA (miR)-383-5p acted as a 'sponge' for lncRNA TMPO-AS1. The expression levels of lncRNA TMPO-AS1 and miR-383-5p were mutually inhibited in pancreatic carcinoma. Moreover, miR-383-5p was revealed to directly target SRY-related high-mobility group box 11 (SOX11). Notably, SOX11 could promote the occurrence of pancreatic carcinoma by interacting with the lncRNA TMPO-AS1/miR-383-5p axis. In conclusion, upregulation of lncRNA TMPO-AS1 promoted tumor growth, cell migration and invasion in pancreatic carcinoma by downregulating miR-383-5p and upregulating SOX11.
Collapse
Affiliation(s)
- Feng Xue
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Xin Song
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Shuai Zhang
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Meiwei Niu
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Yu Cui
- Department of Oncology, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Ye Wang
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| | - Ting Zhao
- Department of General Surgery, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
12
|
Zhao MM, Ge LY, Yang LF, Zheng HX, Chen G, Wu LZ, Shi SM, Wang N, Hang YP. LncRNA NEAT1/ miR-204/ NUAK1 Axis is a Potential Therapeutic Target for Non-Small Cell Lung Cancer. Cancer Manag Res 2020; 12:13357-13368. [PMID: 33402847 PMCID: PMC7778439 DOI: 10.2147/cmar.s277524] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Long non-coding RNA (lncRNA) is a key part of non-coding RNA, and more and more evidence has revealed that it plays a vital role in tumors. NEAT1 is a lncRNA discovered in the early stage. However, it is still unclear whether NEAT1 and miR-204 play a regulatory role in lung cancer (LC). This research aimed to determine the biological function of NEAT1/miR-204 in non-small cell lung cancer (NSCLC). Materials and Methods In order to research the function of NEAT1 in NSCLC, RT-PCR, Western blot, luciferase assay and RNA immunoprecipitation assay were used to determine the relationship between NEAT1, miR-204 and NUAK1. CCK8 test, cell migration and invasion test were used to explore the influence of NEAT1 on proliferation and metastasis of LC cells. Tumor allotransplantation was used to detect the influence of NEAT1 on the growth of LC. Results The results revealed that NEAT1 was obviously enhanced in LC cell lines. Further functional analysis showed that low expression of NEAT1 obviously suppressed the growth, migration and invasion of NSCLC and facilitated cell apoptosis. Determination of luciferase reporter gene revealed that miR-204 was the direct target of NEAT1 in LC. In addition, NUAK1 was called the direct target of miR-204, and miR-204/NUAK1 had saved the role of NEAT1 in NSCLC cells. Tumor allotransplantation experiments showed that knocking down NEAT1 could inhibit the growth of LC. Conclusion In summary, our results showed that the down-regulation of NEAT1 in NSCLC inhibited its growth, migration and invasion through the miR-204/NUAK1 axis.
Collapse
Affiliation(s)
- Ming-Ming Zhao
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Lin-Yang Ge
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Liang-Feng Yang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Hai-Xia Zheng
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Gang Chen
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Li-Zheng Wu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Shao-Ming Shi
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Nan Wang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| | - Yan-Ping Hang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Gaochun, Nanjing 211300, People's Republic of China
| |
Collapse
|
13
|
Zhou L, Xing C, Zhou D, Yang R, Cai M. Downregulation of lncRNA FGF12-AS2 suppresses the tumorigenesis of NSCLC via sponging miR-188-3p. Open Med (Wars) 2020; 15:986-996. [PMID: 33344773 PMCID: PMC7724005 DOI: 10.1515/med-2020-0219] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Non-small-cell lung carcinoma (NSCLC) seriously threatens the health of human beings. Aberrant expression of lncRNAs has been confirmed to be related with the progression of multiple malignant tumors, including NSCLC. LncRNA FGF12-AS2 has been considered to be upregulated in NSCLC. However, the mechanism by which FGF12-AS2 promotes the tumorigenesis of NSCLC remains elusive. Methods Gene and protein expressions in NSCLC cells were measured by q-PCR and western blot, respectively. CCK-8 and immunofluorescence staining were performed to detect the cell proliferation. Cell apoptosis was tested by flow cytometry. Transwell assay was used to detect the cell migration and invasion. Finally, the dual luciferase report assay was used to verify the relation among FGF12-AS2, miR-188-3p, and NCAPG2. Results Downregulation of FGF12-AS2 significantly inhibited the proliferation of NSCLC cells via inducing apoptosis. In addition, FGF12-AS2 silencing notably suppressed the migration and invasion of A549 cells. Meanwhile, FGF12-AS2 modulated the progression of NSCLC via regulation of miR-188-3p/NCAPG2 axis. Finally, knockdown of FGF12-AS2 inhibited the tumorigenesis of NSCLC via suppressing the EMT process of NSCLC. Conclusion Downregulation of lncRNA FGF12-AS2 suppressed the tumorigenesis of NSCLC via sponging miR-188-3p. Thus, FGF12-AS2 may serve as a potential target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lili Zhou
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Chen Xing
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Dongxia Zhou
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Rong Yang
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| | - Maohuai Cai
- Department of Oncology, Yancheng Second People’s Hospital, No. 135 Kaifang Avenue, Yancheng 224003, Jiangsu, China
| |
Collapse
|
14
|
Liu G, Yang H, Cao L, Han K, Li G. LncRNA TMPO-AS1 Promotes Proliferation and Invasion by Sponging miR-383-5p in Glioma Cells. Cancer Manag Res 2020; 12:12001-12009. [PMID: 33262650 PMCID: PMC7696628 DOI: 10.2147/cmar.s282539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 01/15/2023] Open
Abstract
Purpose Glioma is one of the most common malignant tumors affecting human health. Long non-coding RNA (lncRNA) TMPO-AS1 participates in the pathogenesis of various cancers. However, the role of lncRNA TMPO-AS1 in glioma remains largely unknown. This study aims to uncover the role of TMPO-AS1 and explore its potential mechanism in glioma. Methods Expression levels of TMPO-AS1 and miR-383-5p in glioma cell lines were measured by real-time quantitative PCR (RT-qPCR). CCK-8, colony formation, wound-healing, and Transwell assays were conducted to determine cell proliferation, migration and invasion abilities, respectively. Western blotting was applied to detect the expression of corresponding proteins. Immunofluorescence assay was performed to measure the expression of Ki67. The binding condition between TMPO-AS1 and miR-383-5p was verified by dual-luciferase reporter assay. Results We found that TMPO-AS1 was up-regulated while miR-383-5p was down-regulated in glioma cell lines, and knockdown of TMPO-AS1 significantly suppressed glioma cell proliferation, migration and invasion abilities. miR-383-5p was demonstrated to be a direct target of TMPO-AS1. Besides, inhibition of miR-383-5p abolished the effects of TMPO-AS1 knockdown on glioma cells. Conclusion In summary, our study revealed that inhibition of lncRNA TMPO-AS1 could suppress glioma progression through targeting miR-383-5p. TMPO-AS1 might be used as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Guoyuan Liu
- Department of Neurosurgery, The People's Hospital of Jimo, Qingdao, Shandong 266000, People's Republic of China
| | - Haiying Yang
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, People's Republic of China
| | - Lei Cao
- Department of Neurosurgery, The People's Hospital of Jimo, Qingdao, Shandong 266000, People's Republic of China
| | - Kun Han
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, People's Republic of China
| | - Guobin Li
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, People's Republic of China
| |
Collapse
|
15
|
Shi W, Song J, Gao Z, Liu X, Wang W. Downregulation of miR-7-5p Inhibits the Tumorigenesis of Esophagus Cancer via Targeting KLF4. Onco Targets Ther 2020; 13:9443-9453. [PMID: 33061430 PMCID: PMC7522318 DOI: 10.2147/ott.s251508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023] Open
Abstract
Background Esophageal cancer (EC) is one of the aggressive gastrointestinal malignancies. It has been reported that microRNAs (miRNAs) play key roles during the tumorigenesis of EC. To identify novel potential targets for EC, differential expressed miRNAs (DEG) between EC and adjacent normal tissues were analyzed with bioinformatics tool. Methods The differential expression of miRNAs between EC and adjacent normal tissues was analyzed. CCK-8 and Ki67 staining were used to detect the cell proliferation. Flow cytometry was performed to test the cell apoptosis. The correlation between miR-7-5p and KLF4 was detected by dual-luciferase report assay. Gene and protein expression in EC cells or in tissues were measured by qRT-PCR and Western blot, respectively. Cell migration and invasion were detected with transwell assay. Xenograft mice model was established to investigate the role of miR-7-5p in EC tumorigenesis in vivo. Results MiR-7-5p was found to be negatively correlated with the survival rate of patient with EC. In addition, downregulation of miR-7-5p significantly inhibited the growth and invasion of EC cells. Meanwhile, miR-7-5p directly targeted KLF4 in EC cells. Moreover, downregulation of miR-7-5p inhibited the tumorigenesis of EC via inactivating MAPK signaling pathway in vivo. Conclusion Downregulation of miR-7-5p notably suppressed the progression of EC via targeting KLF4. Thus, miR-7-5p might serve as a new target for the treatment of EC.
Collapse
Affiliation(s)
- Woda Shi
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, People's Republic of China
| | - Jianxiang Song
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, People's Republic of China
| | - Zhengya Gao
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, People's Republic of China
| | - Xingchen Liu
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, People's Republic of China
| | - Wencai Wang
- Department of Cardio-Thoracic Surgery, Yancheng Third People's Hospital, Yancheng, Jiangsu 224000, People's Republic of China
| |
Collapse
|
16
|
BC032913 as a Novel Antisense Non-coding RNA is Downregulated in Gastric Cancer. J Gastrointest Cancer 2020; 52:928-931. [DOI: 10.1007/s12029-020-00517-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Riahi A, Hosseinpour-Feizi M, Rajabi A, Akbarzadeh M, Montazeri V, Safaralizadeh R. Overexpression of long non-coding RNA MCM3AP-AS1 in breast cancer tissues compared to adjacent non-tumour tissues. Br J Biomed Sci 2020; 78:53-57. [PMID: 32678686 DOI: 10.1080/09674845.2020.1798058] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Altered expression of several long non-coding RNAs (lncRNAs) has been described in numerous malignancies, including breast cancer, and some may have a role in carcinogenesis. We hypothesised differences in the expression of lncRNA MCM3AP-AS1 in breast cancer tissues compared to nearby healthy tissues and potential links with clinical features. METHODS We tested our hypothesis in 102 pairs of breast cancer tumours and adjacent non-tumour tissues from female patients. After RNA extraction, cDNA synthesis was performed for all specimens. The differential gene expression was assessed using Quantitative Real-Time PCR Technique. RESULTS There was a significant overexpression of the lncRNAs in tumour tissues as compared with their adjacent non-tumour tissues (P < 0.001). Expression was significantly linked with the tumour oestrogen receptor expression (P = 0.023) and tumour progesterone receptor expression (P < 0.001). ROC analysis showed an AUC of 0.67 (95% CI 0.60-0.75) (P < 0.001) with sensitivity and specificity of 58% and 76%, respectively. CONCLUSION The lncRNA MCM3AP-AS1 may be a novel breast cancer lncRNA with high expression levels in breast cancer patients' tissue. Further investigations are needed to confirm its uses as a potential molecular marker and therapeutic target.
Collapse
Affiliation(s)
- A Riahi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz , Tabriz, Iran
| | - M Hosseinpour-Feizi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz , Tabriz, Iran
| | - A Rajabi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz , Tabriz, Iran
| | - M Akbarzadeh
- Department of Biology, Faculty of Sciences, Azerbaijan Shahid Madani University , Tabriz, Iran
| | - V Montazeri
- Department of Thoracic Surgery, Faculty of Medicine, Tabriz University of Medical Sciences\Surgery Ward, Nour-Nejat Hospital , Tabriz, Iran
| | - R Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz , Tabriz, Iran
| |
Collapse
|
18
|
Zhao L, Li Y, Song A. Inhibition of lncRNA TMPO‑AS1 suppresses proliferation, migration and invasion of colorectal cancer cells by targeting miR‑143‑3p. Mol Med Rep 2020; 22:3245-3254. [PMID: 32945436 PMCID: PMC7453500 DOI: 10.3892/mmr.2020.11427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are widely studied in cancer pathogenesis. Accumulating evidence has demonstrated that lncRNAs are involved in the cellular progression of colorectal cancer (CRC). However, the regulatory mechanism of lncRNA TMPO-antisense (AS)1 in CRC has not been fully elucidated. The present study aimed to elucidate the role and regulatory mechanisms of lncRNA TMPO-AS1 in CRC. In the present study, the expression levels of TMPO-AS1 and microRNA-143-3p (miR-143-3p) were detected using reverse transcription-quantitative PCR assay. The relative protein expression levels were measured via western blot analysis. MTT and Transwell assays were used to determine cell proliferation, migration and invasion, while a luciferase reporter assay was performed to assess the relationship between TMPO-AS1 and miR-143-3p. In addition, a tumor animal model was used to investigate the effect of TMPO-AS1 on tumor growth in CRC in vivo. TMPO-AS1 expression was increased and miR-143-3p expression was decreased in CRC cells. TMPO-AS1 knockdown and miR-143-3p overexpression significantly inhibited cell proliferation, migration and invasion of CRC cells. Luciferase reporter assay results demonstrated that miR-143-3p was a direct target of TMPO-AS1. Inhibition of miR-143-3p could alleviate the suppressive effects of TMPO-AS1 deletion on cell proliferation, migration and invasion of CRC cells. Furthermore, TMPO-AS1 deletion could inhibit tumor growth in CRC in vivo. It was concluded that TMPO-AS1 regulated cell proliferation, migration and invasion of CRC cells by targeting miR-143-3p. These findings provided a new regulatory network and therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Lei Zhao
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 733000, P.R. China
| | - Yu Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 733000, P.R. China
| | - Ailin Song
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 733000, P.R. China
| |
Collapse
|
19
|
Guo X, Wang Y. LncRNA TMPO-AS1 promotes hepatocellular carcinoma cell proliferation, migration and invasion through sponging miR-329-3p to stimulate FOXK1-mediated AKT/mTOR signaling pathway. Cancer Med 2020; 9:5235-5246. [PMID: 32462698 PMCID: PMC7367632 DOI: 10.1002/cam4.3046] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 01/27/2023] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is one of the leading causes of cancer‐related death worldwide. Numerous analyses have revealed the abnormal expression of long non‐coding RNAs (lncRNAs) in HCC cells. This study aims to explore biological functions of lncRNA TMPO‐AS1 (TMPO antisense RNA 1) in HCC cell proliferation, apoptosis, invasion and migration. Methods The gene expression in HCC tissues and cell lines were measured by qRT‐PCR. The role of TMPO‐AS1 in HCC was confirmed by CCK‐8, colony formation, TUNEL, transwell and western blot as well as by in vivo experiments. RNA pull down and luciferase reporter assays were utilized to prove the binding relationship between TMPO‐AS1/FOXK1 (forkhead box K1) andmiR‐329‐3p. Rescue assays elucidated the regulatory effects of TMPO‐AS1/miR‐329‐3p/FOXK1/AKT/mTOR pathway on cellular activities in HCC. Results TMPO‐AS1was upregulated in HCC tissues and cells and its depletion inhibits HCC cell proliferation, invasion, migration, and EMT process as well as tumor growth. Furthermore, TMPO‐AS1 could bind with miR‐329‐3p, which suppressed HCC cell proliferation. FOXK1 served as the target gene of miR‐329‐3p and TMPO‐AS1 upregulated FOXK1 by sponging miR‐329‐3p in HCC cells. Additionally, FOXK1 overexpression or miR‐329‐3p inhibitor neutralized the repressing effects of TMPO‐AS1 knockdown on HCC development. Finally, it verified that TMPO‐AS1 could regulate AKT/mTOR pathway via FOXK1 to promote HCC. Conclusion TMPO‐AS1 contributes to HCC progression by sponging miR‐329‐3p to activate FOXK1‐mediated AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaobo Guo
- Department of Hematology, Xi'an Central Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yun Wang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|